EP2805732B1 - Conjugués médicament-polymère pour le traitement de l'amyloïdose - Google Patents

Conjugués médicament-polymère pour le traitement de l'amyloïdose Download PDF

Info

Publication number
EP2805732B1
EP2805732B1 EP13382184.3A EP13382184A EP2805732B1 EP 2805732 B1 EP2805732 B1 EP 2805732B1 EP 13382184 A EP13382184 A EP 13382184A EP 2805732 B1 EP2805732 B1 EP 2805732B1
Authority
EP
European Patent Office
Prior art keywords
polymer
drug
linked
spacer
pga
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Not-in-force
Application number
EP13382184.3A
Other languages
German (de)
English (en)
Other versions
EP2805732A1 (fr
Inventor
María Jesús VICENT DOCÓN
Inmaculada Conejos Sanchez
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
Fundacion de la Comunidad Valenciana Centro de Investigacion Principe Felipe
Original Assignee
Fundacion de la Comunidad Valenciana Centro de Investigacion Principe Felipe
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Fundacion de la Comunidad Valenciana Centro de Investigacion Principe Felipe filed Critical Fundacion de la Comunidad Valenciana Centro de Investigacion Principe Felipe
Priority to ES13382184.3T priority Critical patent/ES2611185T3/es
Priority to EP13382184.3A priority patent/EP2805732B1/fr
Priority to PCT/EP2014/058856 priority patent/WO2014187654A1/fr
Priority to US14/892,482 priority patent/US20160114055A1/en
Publication of EP2805732A1 publication Critical patent/EP2805732A1/fr
Application granted granted Critical
Publication of EP2805732B1 publication Critical patent/EP2805732B1/fr
Not-in-force legal-status Critical Current
Anticipated expiration legal-status Critical

Links

Images

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/65Tetracyclines
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K47/00Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient
    • A61K47/50Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates
    • A61K47/51Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent
    • A61K47/62Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent the modifying agent being a protein, peptide or polyamino acid
    • A61K47/64Drug-peptide, drug-protein or drug-polyamino acid conjugates, i.e. the modifying agent being a peptide, protein or polyamino acid which is covalently bonded or complexed to a therapeutically active agent
    • A61K47/645Polycationic or polyanionic oligopeptides, polypeptides or polyamino acids, e.g. polylysine, polyarginine, polyglutamic acid or peptide TAT
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K49/00Preparations for testing in vivo
    • A61K49/001Preparation for luminescence or biological staining
    • A61K49/0013Luminescence
    • A61K49/0017Fluorescence in vivo
    • A61K49/0019Fluorescence in vivo characterised by the fluorescent group, e.g. oligomeric, polymeric or dendritic molecules
    • A61K49/0021Fluorescence in vivo characterised by the fluorescent group, e.g. oligomeric, polymeric or dendritic molecules the fluorescent group being a small organic molecule
    • A61K49/0032Methine dyes, e.g. cyanine dyes
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K49/00Preparations for testing in vivo
    • A61K49/001Preparation for luminescence or biological staining
    • A61K49/0013Luminescence
    • A61K49/0017Fluorescence in vivo
    • A61K49/005Fluorescence in vivo characterised by the carrier molecule carrying the fluorescent agent
    • A61K49/0056Peptides, proteins, polyamino acids
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K49/00Preparations for testing in vivo
    • A61K49/06Nuclear magnetic resonance [NMR] contrast preparations; Magnetic resonance imaging [MRI] contrast preparations
    • A61K49/08Nuclear magnetic resonance [NMR] contrast preparations; Magnetic resonance imaging [MRI] contrast preparations characterised by the carrier
    • A61K49/10Organic compounds
    • A61K49/14Peptides, e.g. proteins
    • A61K49/146Peptides, e.g. proteins the peptide being a polyamino acid, e.g. poly-lysine
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K51/00Preparations containing radioactive substances for use in therapy or testing in vivo
    • A61K51/02Preparations containing radioactive substances for use in therapy or testing in vivo characterised by the carrier, i.e. characterised by the agent or material covalently linked or complexing the radioactive nucleus
    • A61K51/04Organic compounds
    • A61K51/08Peptides, e.g. proteins, carriers being peptides, polyamino acids, proteins
    • A61K51/088Peptides, e.g. proteins, carriers being peptides, polyamino acids, proteins conjugates with carriers being peptides, polyamino acids or proteins
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system
    • A61P25/28Drugs for disorders of the nervous system for treating neurodegenerative disorders of the central nervous system, e.g. nootropic agents, cognition enhancers, drugs for treating Alzheimer's disease or other forms of dementia

Definitions

  • the present invention relates to novel chemical conjugates as well as pharmaceutical compositions containing them for use in therapy and diagnosis and, more particularly, but not exclusively, to novel conjugates of polymers having attached thereto a targeting moiety and one or more therapeutic agents for the treatment of amyloidosis.
  • the present invention is referred to novel polymeric conjugates which is linked at least to a fibril disruptor agent and/or a blocking agent of aggregates in addition of an optional targeting moiety and/or a probe for diagnosis and therapy.
  • the present invention relates to a polymer-drug conjugate, where the polymeric platform transports at least one bioactive agent, selected from the group of anthracyclines antibiotics such as tetracycline, rolitetracycline, minocycline and/or doxycycline and their derivatives, able to disaggregate or break the amyloid fibrils and/or block the aggregates.
  • This conjugate could contain in its structure one or several targeting moieties, which will provide an effective targeting of the polymer-drug conjugate to the selected area for drug activity, increasing the therapeutic efficacy in the treatment of amyloidosis.
  • Amyloidosis is a group of diseases characterized by fibrillar protein deposition in the extracellular space named amyloid, of which Familial Amyloid Polyneuropathy (FAP) and Alzheimer's Disease (AD) are examples.
  • FAP Familial Amyloid Polyneuropathy
  • AD Alzheimer's Disease
  • Familial Amyloidotic Polyneuropathy is a neurodegenerative disorder characterised by systemic extracellular deposition of transthyretin (TTR) amyloid fibrils in several organs, mainly in the peripheral nervous system. This disease is characterised by an ascending sensorimotor polyneuropathy and progressive dysautonomia, becoming usually fatal 10 to 15 years after its onset. TTR, a homotetramer mainly synthesised in the liver and choroid plexus, is responsible for the transport of thyroxine and retinol. Over 100 point mutations have been identified in TTR and associated with FAP; this disease presents its largest focus in Portugal and thus, represents an important topic of research.
  • TTR transthyretin
  • doxycycline is the most advanced fibril disrupter agent under study, being the most effective compound at disaggregating TTR mature fibrils. Doxy was able to disrupt amyloid fibrils in vitro, in the treated animals and also achieved other improvements regarding amyloid markers [2, 4].
  • TRCA Tauroursodeoxycholic Acid
  • TUDCA is a biliary acid with antiapoptotic and antioxidant activity.
  • doxycycline administered to mice with amyloid deposition was more effective than either parent drugs per separate.
  • Significantly lowering of TTR deposition and associated tissue markers were observed.
  • the observed synergistic effect of doxy/TUDCA in the range of human tolerable quantities, in the transgenic TTR mice models prompts their application in FAP, particularly in the early stages of disease.
  • FAP farnesothelial growth factor
  • TTR binds A-beta protein, which plays the major role in the Alzheimer's Disease (AD). It can act like a "chaperone” by preventing formation of A-beta amyloid aggregates and thereby it may halt progression of AD [14].
  • AD Alzheimer's Disease
  • This disorder also an amyloidotic disease, affects 5% of the population over the age of 65 years and 20% over 80 years of age.
  • the present invention provides a response to this necessity with novel polymer-drug conjugates able to enhance the activity already found with doxycycline in the treatment of diseases involving amyloidosis, e.g. FAP, showing better specificity and reduced systemic toxicity.
  • novel polymer-drug conjugates able to enhance the activity already found with doxycycline in the treatment of diseases involving amyloidosis, e.g. FAP, showing better specificity and reduced systemic toxicity.
  • Polymer Therapeutics, first polymeric nanomedicines [15, 16], are the basis for the development of the invention which is based on conjugation of the aforementioned compounds (directly or through a linker to a polymeric carrier) by covalent binding in order to obtain novel polymer-drug conjugates with therapeutic potential to treat these neuropathic disorders for the first time in the field.
  • Conjugation procedures have demonstrated greater specificity and increased or retained activity of the parent molecules, importantly, polymer multivalence allows to achieve synergistic effect with combination therapy [17-19]). Conjugation can also provide improvements on drug stability and can reduce drug toxicity yielding a better therapeutic value.
  • Polymer Therapeutics are well-known as effective drug delivery systems with demonstrated clinical benefits since the 90's [20, 21].
  • polymer-drug conjugates are considered new chemical entities (NCEs) capable to improve bioactive compound properties (changing its pharmacokinetics at whole body (EPR effect) and at cellular level (endocytosis) allowing even to overcome cellular mechanisms of resistance [15, 22]) and decreasing their inherent limitations (short half life, non-specific toxicity, low solubility, poor stability, potential immunogenicity,).
  • one of the main targets of the present invention is the use of advanced nanopharmaceutics based on combination therapy looking at agent synergism. Whilst nanosized systems are well established for the delivery of a single therapeutic agent, only in very recent years has their use been extended to the delivery of multi-agent therapy. In fact, the only example in the clinics comes from a Canadian company, Celator Technologies Inc. who has developed a methodical approach for combination therapy within their liposomal technology [29-33].
  • novel specific nanoconjugates for the treatment of amyloidosis related polyneuropathies are shown in this invention, using as polymeric platform the poly-glutamic acid (PGA) and its derivates.
  • PGA poly-glutamic acid
  • One of the major drawbacks that hinders peptides/proteins drugs application in therapies is their variable solubility, low bioavailability and limited stability, therefore one purpose of the present invention is to move a step further this therapy enhancing the activity already found with Doxy in FAP and AD with polymer-drug conjugates, increasing their specificity and diminishing their systemic toxicity.
  • These vehicles present advantages not just for peptides but also for small chemical compounds, allowing the control of their release at the desired site.
  • the property of polymer multivalency allows the conjugation of several active compounds within the same polymeric carrier, the combination of doxy and other drug in the same polymer carrier would synergistically enhance the therapeutic value of these prodrugs. Synergy could also be effective when conjugates of each drug are administered per separate [18]. Due to the molecular complexity of FAP and AD it is expected to achieve better therapeutic output if more than one molecular pathway of disease is targeted.
  • the present invention in some embodiments thereof, relates to novel chemical conjugates and to uses thereof in therapy and diagnosis and, more particularly, but not exclusively, to novel conjugates of polymers having attached thereto one or more therapeutic agents and optionally a targeting moiety and/or a labelling probe, for treatment and diagnosis of amyloidosis related diseases.
  • the present invention relates to a polymer-drug conjugate where the polymeric backbone bears, at least, a fibrillar disrupting agent selected from the group including: anthracyclines antibiotics like the tetracycline, rolitetracycline, minocycline and /or doxycycline or their derivates, etc,...able to disaggregate or break the amyloid fibrils and/or block the aggregates.
  • This conjugate may contain also in its structure targeting moieties which allow an effective targeting of the polymer-drug conjugate to the diseased area, providing higher efficacy in the treatment and/or diagnosis of amyloidosis related diseases, including polyneuropathic disorders and neurodegenerative diseases like FAP and AD.
  • Polymer-drug conjugates of the present invention have shown a marked specificity retaining or enhancing the inherent activity of the selected bioactive molecules, allowing, thanks to the multivalency of the polymers, the obtaining of synergistic effects through the combination therapy, higher stability and lower side toxicity of the parent drug achieving a better therapeutic index.
  • the present invention provides a polymer-drug conjugate represented in the general formula I, also represented in Figure 1 :
  • compositions which comprise the conjugates herein described with a pharmaceutically acceptable carrier.
  • the polymer-drug conjugate is for use in a method of treating or diagnosing amyloidosis in a subject in need of thereof, the method comprising administering to the subject a therapeutically effective amount of the polymer-drug conjugate as described herein.
  • the present invention in some embodiments thereof, relates to novel chemical polymer-drug conjugates for use in therapy and, more particularly, but not exclusively, to novel conjugates of polymers having attached thereto a fibril disrupter agent and/or a aggregate-blocking agent and/or a targeting moiety and/or labelling moiety and to uses thereof in treating amyloidosis.
  • the present invention relates to a polymer-drug conjugate where the polymeric backbone bears, at least, a fibrillar disrupting agent able to disaggregate or break the amyloid fibrils and/or block the aggregates.
  • This conjugate may contain also in its structure targeting moieties which allow an effective targeting of the polymer-drug conjugate to the diseased area, providing higher efficacy in the treatment and/or diagnosis of amyloidosis.
  • This invention provides novel polymer-drug conjugates and pharmaceutical compositions which contain them for use as pharmaceutical agents against diseases involving amyloidosis, including amyloidosis related to polyneuropathic disorders and neurodegenerative disorders like FAP and AD.
  • the polymer-drug conjugates where the polymeric backbone bears, at least, a bioactive agent for amyloidosis treatment and at least a targeting moiety and a labelling probe are for use in diagnosis and to perform biodistribution studies of the bioactive agent in animal models with amyloidosis related diseases.
  • the present invention relates to polymer-drug conjugates which comprise a polymeric backbone to which is covalent linked at least a bioactive agent, with the capability of disrupting amyloid fibrils and /or blocking aggregates activity.
  • amyloid disrupter/disrupting agent describes any therapeutic agent that directly disaggregates or breaks the amyloid fibrils and/or their deposits and promotes a concatenate effect improving disease conditions as observed by amyloid markers studies.
  • amyloid disrupting agents include, anthracyclines antibiotics such as tetracycline, rolitetracycline, minocycline, and/or doxycycline and their derivates, being preferable the use of doxycycline, as it is represented in the Fig.2B , and its derivate Doxy.-NH 2 .
  • the phrase "aggregates-blocking agent”, as used herein, describes any therapeutic agent that directly interacts with the misfolded protein in its aggregate stage being able to avoid interaction with the receptor, thus avoiding cell death cascades.
  • the described conjugates comprise a polymeric backbone that represents the total plurality of units of the main chain, from which, some of these units are linked to the first therapeutic agent, optionally other ratio of these units is linked to the second bioactive agent, and optionally other ratio has been labelled with a probe for the monitoring of the conjugate; and optionally some of the units of the polymeric backbone remain free and other may be linked to a targeting moiety to allow the whole system to cross the blood-brain barrier.
  • the bioactive agent or the bioactive agents are linked to the polymeric matrix directly or through a spacer or linker, being this linker biodegradable and selected from the group consisting of a pH-sensitive and an enzymatically-cleavable linker. In the case of enzymatic linkages, these are cleaved as a consequence of the presence of enzymes over-expressed in inflamed areas.
  • the union comprises a direct covalent bonding of the active agent, via amide, ester, acetal, hydrazone or disulphide bond; and it may include any amino acid sequence including those substrates of metalloprotases (i.e. MMP-9 (PVGLIG)), cathepsin B (GPLG; R; PL) and other peptide sequences able to be recognised and cleaved in presence of enzymes located in drug release area.
  • MMP-9 metalloprotases
  • GPLG cathepsin B
  • PL cathepsin B
  • secuences Gly-Gly and Leu-Gly are used in the present invention.
  • the polymeric backbone is derived from a polyglutamic acid (PGA).
  • the polymeric backbone is derived from polyglutames such as block-co-polymers, triblocks, where the other blocks include without limitation polyethylenglycol (PEG), water soluble polyamino acid, polylactic acid (PLA), polylactic-co-glycolic acid (PLGA), poly(D,L-lactide-co-glycolide) (PLA/PLGA), poly(hydroxyalkylmethacrylamide), polyglyceron, polyamidoamine (PAMAM) and polyethyleneimine (PEI), and diferent structures of the polyglutamic acid (PGA) represented in the Fig. 2A , e.g. star PGA, brush PGA,etc.) being preferable the use of poly-L-glutamic acid, poly-D-glutamic acid or poly-D,L-glutamic acid.
  • PEG polyethylenglycol
  • PEG polyethyleng
  • the present patent includes all the possible polymer-drug linkages derived from PGA modification of its pendant chains (e.g. alkyne, azide, tiol, halides, activated esters, activated alcohols, protected amines, maleimide groups, acetals, ethylene glycol (EG) of several molecular weights including polyethyleneglycol (PEG from 100 to 20000g/mol) as well as drug modification for achieving all the correspondent type of bond with the polymer chain, directly or through spacers. All the possible combinations are included in this invention.
  • PGA modification of its pendant chains e.g. alkyne, azide, tiol, halides, activated esters, activated alcohols, protected amines, maleimide groups, acetals, ethylene glycol (EG) of several molecular weights including polyethyleneglycol (PEG from 100 to 20000g/mol) as well as drug modification for achieving all the correspondent type of bond with the polymer chain, directly or through space
  • the polymer-drug conjugate of the present invention could contain a targeting moiety able to cross specific biological barriers, for example the blood-brain barrier (BBB).
  • BBB blood-brain barrier
  • This targeting residue can be selected from the group which comprises peptide, proteins o a monoclonal antibody.
  • Examples of targeting residues are any ligand capable to target the transferrin receptor (e.g. the transferrin protein (holo- and apo-transferrin), monoclonal antibodies (e.g. the monoclonal (mAb) OX26), the cyclic peptide CRTIGPSVC, etc.) or to target other receptor present in the BBB such as de low density lipoprotein receptor-related protein 1 (LRP-1) like the peptide Angiopep2.
  • LRP-1 de low density lipoprotein receptor-related protein 1
  • a labelling probe such as a near infrared dye for optical imaging, coordination complexes for MRI, and tracers for PET and SPECT studies.
  • a labelling probe such as a near infrared dye for optical imaging, coordination complexes for MRI, and tracers for PET and SPECT studies.
  • Cy5.5 as probe is preferred in the present invention.
  • mol% describes the number of moles of an attached moiety per 1mol of the polymeric conjugate, multiplied by 100.
  • a 1mol% load of a fibril disrupting agent describes a polymeric conjugate composed of 100 backbone units, whereby 1 backbone unit has the bioactive agent attached thereto and the other 99 units are either free or have other agents attached thereto.
  • the optimal degree of loading of the therapeutically active agent (or agents) and the targeting moiety for a given conjugate and a given use is determined empirically based on the desired properties of the conjugate (e.g. water solubility, therapeutic efficacy, pharmacokinetic profile, toxicity and dosage requirements), and optionally on the amount of the conjugated moiety that can be attached to a polymeric backbone in a synthetic pathway of choice.
  • the drug loading attached to the water soluble polymer can vary.
  • the polymer-drug conjugates are characterized by a load of the therapeutically active agent or the targeting moiety greater than 0.5 mol%.
  • the amount of fibril disrupter drug conjugated per water-soluble polymer can vary.
  • such a composition may comprise from about 1%, about 2%, about 3%, about 4%, about 5%, about 6%, about 7%, about 8.degree./a, about 9%, or about 10%, about 11%, about 12%, about 13%, about 14%, about 15%, about 16%, about 17%, about 18%, about 19%, about 20%, about 21% about 22%, about 23%, about 24%, to about 25% (w/w) fibril disrupter drug relative to the mass of the conjugate.
  • such a composition may comprise from about 26%, about 27%, about 28%, about 29%, about 30%, about 31% about 32%, about 33%, about 34%, about 35%, about 36%, about 37%, about 38%, about 39%, to about 40% or more (w/w) fibril disrupter drug relative to the mass of the conjugate.
  • the present invention provides a polymer-drug conjugate represented in the general formula I, also represented in Figure 1 :
  • R2, R3 and R5 can be used for conjugation of bioactive agents (including low molecular weight drugs, peptides, proteins, antibodies), near infrared dyes, coordination complexes for MRI, PET and SPECT tracers.
  • bioactive agents including low molecular weight drugs, peptides, proteins, antibodies
  • near infrared dyes including near infrared dyes, coordination complexes for MRI, PET and SPECT tracers.
  • the compounds of the present invention may contain one or more basic nitrogen atoms and, therefore they may form salts with acids that also form part of this invention.
  • pharmaceutically acceptable salts include, among others, addition salts with inorganic acids such as hydrochloric, hydrobromic, hydroiodic, nitric, perchloric, sulphuric and phosphoric acid, as well as addition of organic acids as acetic, methanesulfonic, trifluoromethanesulfonic, ethanesulfonic, benzenesulfonic, p-toluenesulfonic, benzoic, camphorsulfonic, mandelic, oxalic, succinic, fumaric, tartaric, and maleic acid.
  • compounds of the present invention may contain one or more acid protons and, therefore, they may form salts with bases, which also form part of this invention.
  • these salts include salts with metal cations, such as for example an alkaline metal ion, an alkaline-earth metal ion or an aluminium ion; or it may be coordinated with an organic with an organic or inorganic base.
  • An acceptable organic base includes among others diethylamine and triethylamine.
  • An acceptable inorganic base includes aluminium hydroxide, calcium hydroxide, potassium hydroxide, sodium carbonate, and sodium hydroxide.
  • Salts derived from pharmaceutically acceptable organic nontoxic bases include salts of primary, secondary, and tertiary amines, substituted amines including naturally occurring substituted amines, cyclic amines, and basic ion exchange resins, such as arginine, betaine, caffeine, choline, N,N-dibenzylethylenediamine, diethylamine, 2-diethylaminoethanol, 2-dimethylaminoethanol, ethanolamine, ethylenediamine, N-ethylmorpholine, ethylpiperidine, glucamine, glucosamine, histidine, hydrabamine, isopropylamine, lysine, methylglucamine, morpholine, piperazine, piperidine, polyamine resins, procaine, purines, theobromine, triethylamine, trimethylamine, tripropylamine, tromethamine, and similar ones.
  • basic ion exchange resins such as arginine
  • Salts can be synthesized from the parent compound which contains a basic or acidic moiety by conventional chemical methods. In general, such salts can be prepared by reaction of the free acid or base forms of these compounds with a stochiometric amount of the appropriate base or acid in water or in an organic solvent, such as ether, ethyl acetate, ethanol, isopropanol, or acetonitrile or in a mixture of them.
  • Some of the compounds of formula I of the present invention may exist in unsolvated as well as solvated forms such as, for example, hydrates.
  • the present invention encompasses all such above-mentioned forms which are pharmaceutically active.
  • Some of the compounds of general formula I may exhibit polymorphism, encompassing the present invention all the possible polymorphic forms, and mixtures thereof.
  • Various polymorphs may be prepared by crystallization under different conditions or by heating or melting the compound followed by gradual or fast cooling. The presence of polymorphs may be determined by solid NMR spectroscopy, IR spectroscopy, differential scanning calorimetry, powder X-ray diffraction or such other techniques.
  • Another aspect of the present invention relates to a process for the preparation of polymer conjugate compounds of formula I, their derivatives, their analogues, their tautomeric forms, their stereoisomers, their polymorphs or their pharmaceutical acceptable salts and solvates.
  • a pharmaceutical composition comprising, as an active ingredient, the conjugate as described herein and a pharmaceutically acceptable carrier.
  • the composition is being packaged in a packaging material and identified in print, in or on the packaging material for use in the treatment of a medical condition associated with amyloidosis.
  • the polymer-drug conjugate is for use in a method of treating amyloidosis in a subject in need of thereof, the method comprising administering to the subject a therapeutically effective amount of the conjugate as described herein.
  • the polymer-drug conjugate is for use in a method of monitoring the conjugate within a body of a patient, the method comprising: administering to the patient the conjugate having a labelling agent as described herein attached thereto; and employing an imaging technique for monitoring a distribution of the conjugate within the body.
  • the compounds of the present invention can be administered in the form of any pharmaceutical formulation.
  • the pharmaceutical formulation will depend upon the nature of the active compound and its route of administration. Any route of administration may be used, for example such as oral, buccal, pulmonary, topical, parenteral (including subcutaneous, intramuscular, and intravenous), transdermal, ocular (ophthalmic), inhalation, intranasal, otic, transmucosal, implant or rectal administration. However oral, intranasal or parenteral administration are preferred.
  • Solid compositions for oral administration include among others tablets, granulates and hard gelatin capsules, formulated both as immediate release or modified release formulations.
  • the compounds of the present invention may be incorporated into oral liquid preparations such as emulsions, solutions, dispersions, suspensions, syrups, elixirs or in the form of soft gelatin capsules.
  • oral liquid preparations such as emulsions, solutions, dispersions, suspensions, syrups, elixirs or in the form of soft gelatin capsules.
  • They may contain commonly-used inert diluents, such as purified water, ethanol, sorbitol, glycerol, polyethylene glycols (macrogols) and propylene glycol.
  • Aid compositions can also contain coadjuvants such as wetting, suspending, sweetening, flavouring agents, preservatives, buffers, chelating agents and antioxidants.
  • Injectable preparations for parenteral administration comprise sterile solutions, suspensions or emulsions in oily or aqueous vehicles, and may contain coadjuvants, such as suspending, stabilizing, tonicity agents or dispersing agents.
  • the compound can also be formulated for its intranasal application. Formulations include particles, powders, solutions wherein the compound is dispersed or dissolved in suitable excipients.
  • the pharmaceutical composition is in the form of nanospheres, microparticles and nanoparticles.
  • the effective dosage of active ingredient may vary depending on the particular compound administered, the route of administration, the nature and severity of the disease to be treated, as well as the age, the general condition and body weight of the patient, among other factors.
  • a representative example of a suitable dosage range is from about 0.001 to about 100 mg/Kg body weight per day, which can be administered as single or divided doses. However, the dosage administered will be generally left to the discretion of the physician.
  • treatment includes treatment, prevention and management of such condition.
  • pharmaceutically acceptable refers to those compounds, compositions, and/or dosage forms which are, within the scope of medical judgement, suitable for use in contact with the tissues of humans and animals without excessive toxicity, irritation, allergic response, or other problem or complication, commensurate with a reasonable benefit/risk ratio.
  • step (b) is performed subsequent to, concomitant with or prior to (c).
  • At least one of the monomer units terminating by the first or the second reactive group further comprises a linker linking the reactive group to the monomeric unit.
  • Chemicals and solvents are either A.R. grade or purified by standard techniques.
  • SEC Size Exclusion chromatography
  • NMR Nuclear Magnetic Resonance
  • LCMS Liquid Chromatography-Mass Spectrometry
  • TEM Transmission electron microscopy
  • Samples were adsorbed to glow-discharged carbon-coated collodion film supported on 200-mesh copper grids.
  • the grids were washed with deionized water and stained with 1% uranyl acetate solution.
  • the grids were visualized with a Zeiss microscope operated at 60 kV.
  • Dynamic Light Scattering (DLS) Particle size was measured using a Malvern Zetasizer Nano ZS instrument, equipped with a 532nm laser at a fixed scattering angle of 90°.
  • Particle Radious (nm) in solution was determined at 37°C for Doxycycline conjugates and in PBS 0.020M PBS at 25°C for RAGE peptide conjugates.
  • Doxycyline conjugation to PGA has been accomplished through different type of linker chemistry, directly to the polymer main chain as well as through cleavable spacers. Synthesis explained below show examples such as ester and amide bonds.
  • Molar percentage of conjugated drug was varied in all the synthesis described below, in order to obtain a family of conjugates and exploit the different properties (activity, conformation in solution, stability) achieved in each final conjugate. Results vary from 1 to 60w%.
  • the amount of fibril disrupter drug conjugated per water-soluble polymer can vary.
  • such a composition may comprise from about 1%, about 2%, about 3%, about 4%, about 5%, about 6%, about 7%, about 8.degree./a, about 9%, or about 10%, about 11%, about 12%, about 13%, about 14%, about 15%, about 16%, about 17%, about 18%, about 19%, about 20%, about 21% about 22%, about 23%, about 24%, to about 25% (w/w) fibril disrupter drug relative to the mass of the conjugate.
  • such a composition may comprise from about 26%, about 27%, about 28%, about 29%, about 30%, about 31% about 32%, about 33%, about 34%, about 35%, about 36%, about 37%, about 38%, about 39%, to about 40% or more (w/w) fibril disrupter drug relative to the mass of the conjugate.
  • doxy 512.94g/mol
  • a catalytic amount of DMAP was added and pH was adjusted to 8 with DIEA. After 12-16h under agitation at room temperature, DMF was evaporated under high vacuum until product was dried.
  • Non-reacted drug as well as reaction organic soluble subproducts were removed by washing the crude with a mixture of CHCl 3 :acetone (4:1) and placed in ice for 30min. Thereafter, solvent was removed by centrifugation and the solid residue was dried under vacuum.
  • Non-reacted drug as well as reaction organic soluble subproducts were removed by washing the crude with a mixture of CHCl 3 :acetone (4:1) and placed in ice for 30min. Thereafter, solvent was removed by centrifugation and the solid residue was dried under vacuum.
  • reagents amount was varied.
  • DIC 0.035mmol, 1.5eq per carboxyl group, 0.836g/cm 3 , 126.20g/mol
  • HOBt 0.035mmol, 1.5eq per carboxyl group, 135.10g/mol
  • Z-Gly-Gly-OH (0.079mmol, 266.25/mol) was added to a round bottom flask and dissolved in 0.5mL anhydrous DMF under nitrogen atmosphere and continuous agitation. Afterwards, DMTMM ⁇ BF 4 (0.087mmol, 1.1eq, 328.07g/mol was added dissolved in 0.5mL of anhydrous DMF. After 10minutes, doxy-NH 2 (0.18mmol, 2.2eq, 557.13g/mol) was added dissolved in 2mL of anhydrous DMF. pH was checked to be 8. The reaction was allowed to proceed for 14h stirring at room temperature and protected from light.
  • PGA (52mg, 0.4mmol unit of glutamic acid) was placed in a round bottom flask and dissolved in anhydrous DMF (5mL) under agitation and N 2 atmosphere.
  • Carboxyl pendant groups were activated with DMTMM ⁇ BF 4 (i.e. for 30% activation, 19mg, 0.06mmol), which were added to main reaction previously dissolved in anhydrous DMF.
  • AA-doxy-NH 2 was added already dissolved in anhydrous DMF. pH was assessed to be 8. Reaction was left under agitation for 24h protected from light at room temperature. Afterwards, solvent was removed by evaporation and residue was washed with MeOH at 4°C. After remove the supernatant by centrifugation, solid product was dried. Yield: 50%.
  • PGA-X-Doxy encompasses all doxycycline conjugates, independently of the type of linkage/spacer between the drug and the polymeric carrier.
  • polymer conjugate salt form NaHCO 3 1M
  • PGA-X-Doxy encompasses all doxycycline conjugates, independently of the type of linkage/spacer between the drug and the polymeric carrier.
  • Conformation in solution is a useful tool for explaining conjugates activity either in vitro or in vivo. Drug availability in the adopted conformation might be directly related with the observed activity.
  • TEM Transmission Electron Microscopy
  • Erythrocytes were isolated by centrifugation at 3000rpm for 10min at 4°C. The plasma supernatant was discarded and the RBCs were washed three times using phosphate buffer saline solution. After the third wash, the RBC pellet was equally divided into two and resuspended in PBS solutions with the appropriate pH values (i.e. 7.4, 6.5 and 5.5). From each RBCs solution, it was added (100uL) to the previously prepared microtitre plates containing the test compounds. As 100% reference of lysis, Tritron X-1%w/v was used as well as PBS as reference. Plates were incubated for 16h at 37°C.
  • H b release was measured spectrophotometrically (OD 570 ). H b release for each sample was expressed as percentage of the release produced by Triton X. PEI and dextran represented the positive and the negative control, respectively. All haemolysis experiments were carried out per triplicate.
  • Example of the conjugate PGA-CONH-Doxy 17wt% is shown in Figure 18 . As expected, compounds are not haemolytic and therefore appropriate for i.v. administration.
  • conjugates were labelled for posterior biodistribution studies using optical imaging, monitoring in vivo as well as ex vivo fluorescence.
  • mice were injected intravenously a single dose of 100uL of the solution.
  • non-injected mice were used.
  • Animals were anesthetized using 1-3% isofluorane.
  • Five mice were imaged at a time and imaging settings were set depending on the fluorescence of the animals. The light emitted from the labelled conjugate was detected, digitalised and electronically displayed as a pseudocolor overlay onto a grey scale animal image. Fluorescent signals were quantified as Efficiency. Mice were euthanized at 0, 4 and 24h post administration. Blood was collected and major organs (liver, lungs, spleen, kidneys, heart and brain) were dissected from mice and fluorescence images of excised tissues were obtained by ex vivo FLI.
  • Ultraturrax device aprox. 1 min, 13000rpm
  • mice for human V30M TTR in a TTR null background were kindly provided by Professor Suichiro Maeda from Yamanashi University. In previous studies, these animals were previously analysed and ⁇ 60% of the animals over 1 year of age were found to have TTR deposition as amyloid, i.e., Congo red (CR)-positive material [36] having non-fibrillar TTR deposits at younger ages.
  • TTR-non fibrilar deposition in the extracted tissues was evaluated by immunohistochemistry as well as possible toxicity of these deposits (apoptosis).
  • Fig. 22 there is represented semi-quantitatively the results obtained for non-fibrillar TTR deposition.
  • the gastrointestinal (GI) tract is the most affected organ regarding fibrillar deposition.
  • Immunohistochemistry results were mainly similar outcomes although Briefly, Fas plays a key role in apoptotic cell death regulation (induced in this experiment by the non-fibrillar TTR).
  • Fas is a tumour-necrosis receptor that binds the Fas ligand resulting in death-inducing signalling complex ending in the apoptosis cascade.
  • BiP is a molecular chaperon synthesised in higher levels when protein folding is disturbed. Then, the process is directly related to cell stress. When drug stops disease advance, levels of Fas and Bip shoud decrease. Immunostainning experiments of Fas and BiP did not produce any profitable result. Treated animals had the same pattern than control animals with disease.
  • FIG. 22 illustrates some of the results of the experiment. Summarising, the findings indicate that treated animals did not show toxicity attributed to the administered conjugate. Furthermore, all animals treated showed normal patterns of liver regarding tissue morphology than the disease control animals. Although TTR is not affected by TTR deposits in this organ, liver is the main producer of the protein. Conjugate administered did not provoke toxicity in the analysed organs.
  • the illustrated groups in Fig. 22 are:

Landscapes

  • Health & Medical Sciences (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Veterinary Medicine (AREA)
  • Animal Behavior & Ethology (AREA)
  • General Health & Medical Sciences (AREA)
  • Public Health (AREA)
  • Epidemiology (AREA)
  • Proteomics, Peptides & Aminoacids (AREA)
  • Engineering & Computer Science (AREA)
  • Chemical & Material Sciences (AREA)
  • Medicinal Chemistry (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Biomedical Technology (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • Physics & Mathematics (AREA)
  • Optics & Photonics (AREA)
  • Neurology (AREA)
  • Neurosurgery (AREA)
  • Molecular Biology (AREA)
  • Radiology & Medical Imaging (AREA)
  • Hospice & Palliative Care (AREA)
  • General Chemical & Material Sciences (AREA)
  • Organic Chemistry (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • Psychiatry (AREA)
  • Pharmaceuticals Containing Other Organic And Inorganic Compounds (AREA)
  • Medicines Containing Antibodies Or Antigens For Use As Internal Diagnostic Agents (AREA)

Claims (8)

  1. Composé de conjugué de médicament-polymère de formule générale I,
    Figure imgb0005
    où le squelette polymérique comporte, au moins, un agent bioactif, et optionnellement un deuxième agent bioactif, optionnellement une sonde pour la surveillance du conjugué et optionnellement un fragment de ciblage, et
    Dans lequel :
    R1 représente
    un groupe alkyle, un point de liaison C-terminal défini (alcyne, azoture, thiol, thiols activés, halogénures, alcènes, esters activés, alcools activés, amines protégés, groupe maléimide, acétals, groupes carboxyliques activés), l'éthylène glycol (EG) de poids moléculaires différents y compris le poly(éthylène glycol) (PEG de 100 à 20000 g/mol).
    R2 représente un atome d'hydrogène, un groupe alkyle, un point de liaison C-terminal défini (alcyne, azoture, thiols, thiols activés, halogénures, alcènes, esters activés, alcools activés, amines protégés, groupe maléimide, acétals, groupes carboxyliques activés), l'éthylène glycol (EG) de poids moléculaires différents y compris le poly(éthylène glycol) (PEG de 100 à 20000 g/mol), le PEG-thiol, le PEG-4TP.
    R3 représente l'espaceur de liaison ou le lien entre la chaîne principale polymère et l'agent bioactif (R4) lui-même ou dérivé, et est un groupe alkyle, un point de liaison C-terminal défini (alcyne, azoture, thiol, thiols activés, halogénures, alcènes, esters activés, alcools activés, amines protégés, groupe maléimide, acétals, groupes carboxyliques activés), l'éthylène glycol (EG) de poids moléculaires différents y compris le poly(éthylène glycol) (PEG de n=2-16), des aminoacides tels que la lysine, l'arginine, l'imidazole, l'histidine, la cystéine et des groupes amino secondaires et tertiaires et des séquences aminoacides et des séquences aminoacides.
    R4 est le médicament choisi pour le traitement de l'amyloïdose, choisi dans le groupe qui comprend les antibiotiques anthracyclines tels que la tétracycline, la rolitétracycline, la minocycline, la doxycycline et leurs dérivés, et le médicament peut être covalent lié à la chaîne de polymère elle-même ou préalablement dérivatisée (y compris l'introduction d'amine, de thiol, de carbonyle, de vinyle, d'alcool ou de groupes carboxyles)
    R5 représente l'espaceur de liaison ou lien entre la chaîne principale polymère et l'agent bioactif R6 lui-même ou dérivatisé et est un groupe alkyle, un point de liaison C-terminal défini (alcyne, azoture, thiol, thiols activés, halogénures, alcènes, esters activés, alcools activés, amines protégés, groupe maléimide, acétals, groupes carboxyliques activés), l'éthylène glycol (EG) de poids moléculaires différents y compris le poly(éthylène glycol) (PEG de n=2 à n=16), des aminoacides tels que la lysine, l'arginine, l'imidazole, l'histidine, la cystéine et des groupes amino secondaires et tertiaires et des séquences aminoacides.
    R6 est le deuxième médicament choisi pour le traitement de l'amyloïdose, choisi dans le groupe qui comprend les antibiotiques anthracyclines tels que la tétracycline, la rolitétracycline, la minocycline, la doxycycline et leurs dérivés (le médicament peut être covalent lié à la chaîne de polymère elle-même ou préalablement dérivée) ; ou un fragment d'étiquetage exploité pour la surveillance du conjugué, pour des expériences de biodistribution ou comme sonde diagnostic où l'agent d'étiquetage comprend des sondes fluorescentes pour l'imagerie optique telles que Cy5.5, des complexes de coordination pour l'IRM, ou des traceurs pour la TEP et la TEMP, y compris les agents de chélation DTPA, DOTA, NOTA, NODA et des liants métalliques tels que le gallium, le technétium, le gadolinium, l'indium.
    x est l'unité monomère comprise dans R1, de 1 à 1000.
    y est un entier ayant une valeur telle que y/(x+y+z+p+q) multiplié par 100 est dans la plage de 0,01 à 99,9.
    z est un entier ayant une valeur telle que z/(x+y+z+p+q) multiplié par 100 est dans la plage de 0,01 à 99,9.
    p est un entier ayant une valeur telle que z/(x+y+z+p+q) multiplié par 100 est dans la plage de 0,01 à 99.9.
    q est l'unité monomère comprise dans R2, de 1 à 1000.
    R2, R3 et R5 peuvent être utilisés pour la conjugaison d'agents bioactifs (y compris les médicaments de faible poids moléculaire, les peptides, les protéines, les anticorps), de colorants dans le proche infrarouge, de complexes de coordination pour l'IRM, la TEP et les traceurs TEMP,
    et/ou leurs sels, polymorphes, solvates et hydrates pour une utilisation dans le traitement ou le diagnostic des maladies associées à l'amyloïdose.
  2. Conjugué de médicament-polymère pour une utilisation selon la revendication 1, dans lequel :
    - Il y a un médicament conjugué dans R4, lié au squelette de polymère directement ou par le biais d'un espaceur (R3),
    - Il y a un médicament conjugué dans R4 (lié au squelette de polymère directement ou par le biais d'un espaceur (R3)) et un fragment de ciblage dans R6 (lié au squelette de polymère directement ou par le biais d'un espaceur) ou dans R2.
    - -Il y a un médicament conjugué dans R4 (lié au squelette de polymère directement ou par le biais d'un espaceur (R3)) ; un fragment de ciblage dans R6 (lié au squelette de polymère directement ou par le biais d'un espaceur) ou dans R2 ; et une sonde d'étiquetage pour le diagnostic dans R6 (lié au squelette de polymère directement ou par le biais d'un espaceur) ou dans R2.
    - -Il y a un médicament conjugué dans R4 (lié au squelette de polymère directement ou par le biais d'un espaceur (R3)) et une sonde d'étiquetage pour le diagnostic dans R6 (lié au squelette de polymère directement ou par le biais d'un espaceur) ou dans R2.
    - Il y a deux médicaments conjugués dans R4 et R6 (liés au squelette de polymère directement ou par le biais d'un espaceur (R3 et R5, respectivement)).
    - Il y a deux médicaments conjugués dans R4 et R6 (liés au squelette de polymère directement ou par le biais d'un espaceur (R3 et R5, respectivement)), un fragment de ciblage dans R6 (lié au squelette de polymère directement ou par le biais d'un espaceur) ou dans R2 ; et une sonde d'étiquetage pour le diagnostic dans R6 (lié au squelette polymère directement ou par le biais d'un espaceur) ou dans R2.
    - Il y a deux médicaments conjugués dans R4 et R6 (liés au squelette de polymère directement ou par le biais d'un espaceur (R3 et R5, respectivement) et une sonde d'étiquetage pour le diagnostic dans R6 (lié au squelette de polymère directement ou par le biais d'un espaceur) ou dans R2.
    - Il y a deux médicaments conjugués dans R4 et R6 (liés au squelette de polymère directement ou par le biais d'un espaceur (R3 et R5, respectivement)) et un fragment de ciblage dans R6 (lié au squelette de polymère directement ou par le biais d'un espaceur) ou dans R2.
  3. Conjugué de médicament-polymère pour une utilisation selon l'une quelconque des revendications 1 et 2, choisi dans le groupe consistant en : PGA-COO-Doxycycline, PGA- CONH-Doxycycline, PGA-Leu Gly-Doxycycline, PGA-Gly-Gly-Doxycycline, PGA- Doxycycline-Cy5.5, PGA-Doxycycline-DOTA/Ga, avec une concentration différente de sonde de médicament/étiquetage dans tous les groupes.
  4. Conjugué de médicament-polymère pour une utilisation selon l'une quelconque des revendications 1 - 3 caractérisé en ce qu'il contient une concentration d'agent bioactif ou de fragment de ciblage supérieure à 0,5 % molaire.
  5. Conjugué de médicament-polymère pour une utilisation selon les revendications 1 - 4, dans lequel le traitement ou le diagnostic de maladies associées à l'amyloïdose est choisi parmi la polyneuropathie amyloïde familiale (PAF) et la maladie d'Alzheimer (MA).
  6. Conjugué de médicament-polymère pour une utilisation selon l'une quelconque des revendications 1 - 5, conjointement à au moins un véhicule pharmaceutiquement acceptable.
  7. Conjugué de médicament-polymère pour une utilisation selon la revendication 6 formulé pour son administration parentérale, orale, topique, nasale et/ou rectale.
  8. Procédé pour obtenir les conjugués de médicament-polymère selon les revendications 1 à 4, qui comprend les étapes suivantes :
    (a) co-polymériser une pluralité d'unités monomériques du polymère, au moins l'une des unités monomériques terminant par un premier groupe réactif, et au moins l'une des unités monomériques terminant par un deuxième groupe réactif, pour obtenir ainsi un copolymère qui comprend une pluralité d'unités de squelette, au moins une unité de squelette ayant le premier groupe réactif et au moins une unité de squelette ayant le deuxième groupe réactif, le premier groupe réactif étant capable de réagir avec le fragment de ciblage et le deuxième réactif étant capable de réagir avec l'agent actif thérapeutiquement, ou :
    (b) Polymérisation d'une seule unité monomérique au moyen d'un polymère bloc comme initiateur, et/ou
    Post-modification après la polymérisation du premier bloc synthétisé polymérique obtenant deux ou plusieurs groupes de fin réactifs dans la chaîne de polymère de début avec des longueurs de polymère modifiables au squelette original, donnant différentes structures polymériques finales et/ou conformations dans la solution, et/ou
    Construction d'une structure à base de polymère tribloc où le bloc au milieu possède le premier groupe réactif étant capable de réagir avec l'agent (les agents) thérapeutique(s) et/ou l'agent d'étiquetage et l'un des blocs polymériques de fin termine par un deuxième groupe réactif étant capable de réagir avec l'agent d'étiquetage, le fragment de ciblage ou un deuxième agent thérapeutique.
    (c) Faire réagir le véhicule polymère avec le fragment de ciblage ou un dérivé de celui-ci, à travers le premier groupe réactif, pour obtenir ainsi un véhicule polymérique ayant le fragment de ciblage attaché à un squelette polymérique de celui-ci ; et
    (d) Faire réagir le véhicule polymère avec l'agent actif thérapeutique ou un dérivé de celui-ci, à travers le deuxième groupe réactif, pour obtenir ainsi le véhicule polymère ayant l'agent actif thérapeutique attaché à un squelette polymérique de celui-ci, obtenant ainsi le conjugué de formule 1 ou faisant réagir le véhicule de polymère avec le premier agent réactif thérapeutiquement ou un dérivé de celui-ci, et deuxièmement le prochain agent thérapeutiquement (déjà lié à un autre polymère ou destiné à être lié au polymère initial post-modifié) à la chaîne de polymère principale.
EP13382184.3A 2013-05-20 2013-05-20 Conjugués médicament-polymère pour le traitement de l'amyloïdose Not-in-force EP2805732B1 (fr)

Priority Applications (4)

Application Number Priority Date Filing Date Title
ES13382184.3T ES2611185T3 (es) 2013-05-20 2013-05-20 Conjugados polímero-fármaco para el tratamiento de la amiloidosis
EP13382184.3A EP2805732B1 (fr) 2013-05-20 2013-05-20 Conjugués médicament-polymère pour le traitement de l'amyloïdose
PCT/EP2014/058856 WO2014187654A1 (fr) 2013-05-20 2014-04-30 Conjugues de medicaments polymeres pour le traitement de l'amylose
US14/892,482 US20160114055A1 (en) 2013-05-20 2014-04-30 Polymer drug conjugates for the treatment of amyloidosis

Applications Claiming Priority (1)

Application Number Priority Date Filing Date Title
EP13382184.3A EP2805732B1 (fr) 2013-05-20 2013-05-20 Conjugués médicament-polymère pour le traitement de l'amyloïdose

Publications (2)

Publication Number Publication Date
EP2805732A1 EP2805732A1 (fr) 2014-11-26
EP2805732B1 true EP2805732B1 (fr) 2016-09-28

Family

ID=48656011

Family Applications (1)

Application Number Title Priority Date Filing Date
EP13382184.3A Not-in-force EP2805732B1 (fr) 2013-05-20 2013-05-20 Conjugués médicament-polymère pour le traitement de l'amyloïdose

Country Status (4)

Country Link
US (1) US20160114055A1 (fr)
EP (1) EP2805732B1 (fr)
ES (1) ES2611185T3 (fr)
WO (1) WO2014187654A1 (fr)

Families Citing this family (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20130065042A1 (en) 2011-03-11 2013-03-14 The Board Of Trustees Of The University Of Illinois Micro-Vascular Materials And Composites For Forming The Materials
WO2020193802A1 (fr) * 2019-03-28 2020-10-01 Fundación De La Comunidad Valenciana Centro De Investigación Príncipe Felipe Conjugués polymères et leurs utilisations

Family Cites Families (5)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US4126680A (en) * 1977-04-27 1978-11-21 Pfizer Inc. Tetracycline antibiotic compositions
US8067006B2 (en) * 2005-04-06 2011-11-29 Immunomedics, Inc. Polymeric carriers of therapeutic agents and recognition moieties for antibody-based targeting of disease sites
ES2296484B1 (es) 2005-11-23 2009-04-01 Fundacion De La Comunidad Valenciana Centro De Investigacion Principe Felipe (90%) Composicion farmaceutica para inhibir la apoptosis.
WO2008124735A2 (fr) * 2007-04-10 2008-10-16 Nitto Denko Corporation Supports de médicaments multifonctionnels à base de polyglutamate
ES2402614B1 (es) * 2011-10-24 2014-03-18 Centro De Investigación Principe Felipe Sintesis controlada de poliglutamatos con baja polidispersidad y arquitecturas versátiles

Also Published As

Publication number Publication date
WO2014187654A1 (fr) 2014-11-27
EP2805732A1 (fr) 2014-11-26
ES2611185T3 (es) 2017-05-05
US20160114055A1 (en) 2016-04-28

Similar Documents

Publication Publication Date Title
Zhou et al. Dual-pH sensitive charge-reversal nanocomplex for tumor-targeted drug delivery with enhanced anticancer activity
Xu et al. Smart and hyper-fast responsive polyprodrug nanoplatform for targeted cancer therapy
JP6585504B2 (ja) ポルフィリン修飾されたテロデンドリマー
Melnyk et al. Therapeutic potential of polypeptide-based conjugates: rational design and analytical tools that can boost clinical translation
Zhu et al. PEGylated PAMAM dendrimer-doxorubicin conjugates: in vitro evaluation and in vivo tumor accumulation
Miyano et al. Anionic amino acid dendrimer− trastuzumab conjugates for specific internalization in HER2-positive cancer cells
Costa et al. Functionalised carbon nanotubes enhance brain delivery of amyloid-targeting Pittsburgh compound B (PiB)-derived ligands
US20130336889A1 (en) Nanoparticle and method for detecting or treating a tumor using the same
Lee et al. Tumor targeting efficiency of bare nanoparticles does not mean the efficacy of loaded anticancer drugs: importance of radionuclide imaging for optimization of highly selective tumor targeting polymeric nanoparticles with or without drug
Shan et al. A paclitaxel-conjugated adenovirus vector for targeted drug delivery for tumor therapy
Wu et al. Albumin-based nanoparticles as methylprednisolone carriers for targeted delivery towards the neonatal Fc receptor in glomerular podocytes
Li et al. Choline derivate-modified doxorubicin loaded micelle for glioma therapy
Wei et al. Enhancing the accumulation of polymer micelles by selectively dilating tumor blood vessels with NO for highly effective cancer treatment
Nabi et al. Mucin-1 conjugated polyamidoamine-based nanoparticles for image-guided delivery of gefitinib to breast cancer
Chandna et al. Targeted proapoptotic anticancer drug delivery system
EP2531221B1 (fr) Macromolécule polyanionique multivalente pour le ciblage intracellulaire de la prolifération et de la synthèse de protéine
CA2957721A1 (fr) Administration d'un dendrimere selectif dans des tumeurs cerebrales
US10709794B2 (en) Photodynamic therapy composition
Conejos-Sánchez et al. Polymer-doxycycline conjugates as fibril disrupters: An approach towards the treatment of a rare amyloidotic disease
Han et al. Acid Active Receptor‐Specific Peptide Ligand for In Vivo Tumor‐Targeted Delivery
Zhang et al. Dual-responsive doxorubicin-loaded nanomicelles for enhanced cancer therapy
Pan et al. Nuclear-targeting TAT-PEG-Asp8-doxorubicin polymeric nanoassembly to overcome drug-resistant colon cancer
Zhao et al. Polyethylenimine-based theranostic nanoplatform for glioma-targeting single-photon emission computed tomography imaging and anticancer drug delivery
US10201622B2 (en) Tumour-targeted theranostic
Zhou et al. A linear polyethylenimine (LPEI) drug conjugate with reversible charge to overcome multidrug resistance in cancer cells

Legal Events

Date Code Title Description
PUAI Public reference made under article 153(3) epc to a published international application that has entered the european phase

Free format text: ORIGINAL CODE: 0009012

17P Request for examination filed

Effective date: 20130520

AK Designated contracting states

Kind code of ref document: A1

Designated state(s): AL AT BE BG CH CY CZ DE DK EE ES FI FR GB GR HR HU IE IS IT LI LT LU LV MC MK MT NL NO PL PT RO RS SE SI SK SM TR

AX Request for extension of the european patent

Extension state: BA ME

R17P Request for examination filed (corrected)

Effective date: 20150525

RBV Designated contracting states (corrected)

Designated state(s): AL AT BE BG CH CY CZ DE DK EE ES FI FR GB GR HR HU IE IS IT LI LT LU LV MC MK MT NL NO PL PT RO RS SE SI SK SM TR

GRAP Despatch of communication of intention to grant a patent

Free format text: ORIGINAL CODE: EPIDOSNIGR1

INTG Intention to grant announced

Effective date: 20160506

GRAS Grant fee paid

Free format text: ORIGINAL CODE: EPIDOSNIGR3

GRAA (expected) grant

Free format text: ORIGINAL CODE: 0009210

AK Designated contracting states

Kind code of ref document: B1

Designated state(s): AL AT BE BG CH CY CZ DE DK EE ES FI FR GB GR HR HU IE IS IT LI LT LU LV MC MK MT NL NO PL PT RO RS SE SI SK SM TR

REG Reference to a national code

Ref country code: GB

Ref legal event code: FG4D

REG Reference to a national code

Ref country code: CH

Ref legal event code: EP

REG Reference to a national code

Ref country code: AT

Ref legal event code: REF

Ref document number: 832184

Country of ref document: AT

Kind code of ref document: T

Effective date: 20161015

REG Reference to a national code

Ref country code: IE

Ref legal event code: FG4D

REG Reference to a national code

Ref country code: DE

Ref legal event code: R096

Ref document number: 602013012128

Country of ref document: DE

REG Reference to a national code

Ref country code: DE

Ref legal event code: R079

Ref document number: 602013012128

Country of ref document: DE

Free format text: PREVIOUS MAIN CLASS: A61K0047480000

Ipc: A61K0047500000

REG Reference to a national code

Ref country code: LT

Ref legal event code: MG4D

PG25 Lapsed in a contracting state [announced via postgrant information from national office to epo]

Ref country code: LT

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20160928

Ref country code: HR

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20160928

Ref country code: RS

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20160928

Ref country code: FI

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20160928

Ref country code: NO

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20161228

REG Reference to a national code

Ref country code: NL

Ref legal event code: MP

Effective date: 20160928

REG Reference to a national code

Ref country code: AT

Ref legal event code: MK05

Ref document number: 832184

Country of ref document: AT

Kind code of ref document: T

Effective date: 20160928

PG25 Lapsed in a contracting state [announced via postgrant information from national office to epo]

Ref country code: LV

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20160928

Ref country code: GR

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20161229

Ref country code: NL

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20160928

Ref country code: SE

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20160928

PG25 Lapsed in a contracting state [announced via postgrant information from national office to epo]

Ref country code: EE

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20160928

Ref country code: RO

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20160928

REG Reference to a national code

Ref country code: ES

Ref legal event code: FG2A

Ref document number: 2611185

Country of ref document: ES

Kind code of ref document: T3

Effective date: 20170505

REG Reference to a national code

Ref country code: FR

Ref legal event code: PLFP

Year of fee payment: 5

PG25 Lapsed in a contracting state [announced via postgrant information from national office to epo]

Ref country code: IS

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20170128

Ref country code: BE

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20160928

Ref country code: BG

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20161228

Ref country code: PT

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20170130

Ref country code: SK

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20160928

Ref country code: PL

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20160928

Ref country code: SM

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20160928

Ref country code: CZ

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20160928

Ref country code: AT

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20160928

REG Reference to a national code

Ref country code: DE

Ref legal event code: R097

Ref document number: 602013012128

Country of ref document: DE

PG25 Lapsed in a contracting state [announced via postgrant information from national office to epo]

Ref country code: IT

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20160928

PG25 Lapsed in a contracting state [announced via postgrant information from national office to epo]

Ref country code: DK

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20160928

PGFP Annual fee paid to national office [announced via postgrant information from national office to epo]

Ref country code: FR

Payment date: 20170525

Year of fee payment: 5

Ref country code: DE

Payment date: 20170530

Year of fee payment: 5

Ref country code: GB

Payment date: 20170530

Year of fee payment: 5

PLBE No opposition filed within time limit

Free format text: ORIGINAL CODE: 0009261

STAA Information on the status of an ep patent application or granted ep patent

Free format text: STATUS: NO OPPOSITION FILED WITHIN TIME LIMIT

PG25 Lapsed in a contracting state [announced via postgrant information from national office to epo]

Ref country code: LU

Free format text: LAPSE BECAUSE OF NON-PAYMENT OF DUE FEES

Effective date: 20170531

PGFP Annual fee paid to national office [announced via postgrant information from national office to epo]

Ref country code: ES

Payment date: 20170601

Year of fee payment: 5

26N No opposition filed

Effective date: 20170629

PG25 Lapsed in a contracting state [announced via postgrant information from national office to epo]

Ref country code: SI

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20160928

REG Reference to a national code

Ref country code: CH

Ref legal event code: PL

PG25 Lapsed in a contracting state [announced via postgrant information from national office to epo]

Ref country code: MC

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20160928

REG Reference to a national code

Ref country code: IE

Ref legal event code: MM4A

PG25 Lapsed in a contracting state [announced via postgrant information from national office to epo]

Ref country code: LI

Free format text: LAPSE BECAUSE OF NON-PAYMENT OF DUE FEES

Effective date: 20170531

Ref country code: CH

Free format text: LAPSE BECAUSE OF NON-PAYMENT OF DUE FEES

Effective date: 20170531

PG25 Lapsed in a contracting state [announced via postgrant information from national office to epo]

Ref country code: LU

Free format text: LAPSE BECAUSE OF NON-PAYMENT OF DUE FEES

Effective date: 20170520

PG25 Lapsed in a contracting state [announced via postgrant information from national office to epo]

Ref country code: IE

Free format text: LAPSE BECAUSE OF NON-PAYMENT OF DUE FEES

Effective date: 20170520

PG25 Lapsed in a contracting state [announced via postgrant information from national office to epo]

Ref country code: MT

Free format text: LAPSE BECAUSE OF NON-PAYMENT OF DUE FEES

Effective date: 20170520

PG25 Lapsed in a contracting state [announced via postgrant information from national office to epo]

Ref country code: AL

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20160928

REG Reference to a national code

Ref country code: DE

Ref legal event code: R119

Ref document number: 602013012128

Country of ref document: DE

GBPC Gb: european patent ceased through non-payment of renewal fee

Effective date: 20180520

PG25 Lapsed in a contracting state [announced via postgrant information from national office to epo]

Ref country code: GB

Free format text: LAPSE BECAUSE OF NON-PAYMENT OF DUE FEES

Effective date: 20180520

Ref country code: DE

Free format text: LAPSE BECAUSE OF NON-PAYMENT OF DUE FEES

Effective date: 20181201

Ref country code: FR

Free format text: LAPSE BECAUSE OF NON-PAYMENT OF DUE FEES

Effective date: 20180531

PG25 Lapsed in a contracting state [announced via postgrant information from national office to epo]

Ref country code: HU

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT; INVALID AB INITIO

Effective date: 20130520

REG Reference to a national code

Ref country code: ES

Ref legal event code: FD2A

Effective date: 20190913

PG25 Lapsed in a contracting state [announced via postgrant information from national office to epo]

Ref country code: CY

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20160928

Ref country code: ES

Free format text: LAPSE BECAUSE OF NON-PAYMENT OF DUE FEES

Effective date: 20180521

PG25 Lapsed in a contracting state [announced via postgrant information from national office to epo]

Ref country code: MK

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20160928

PG25 Lapsed in a contracting state [announced via postgrant information from national office to epo]

Ref country code: TR

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20160928